Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(10)2022 May 14.
Article in English | MEDLINE | ID: mdl-35628302

ABSTRACT

Fibroblast growth factor 21 (FGF21) functions as a polypeptide hormone to regulate glucose and lipid metabolism, and its expression is regulated by cellular metabolic stress. Pyruvate is an important intermediate metabolite that acts as a key hub for cellular fuel metabolism. However, the effect of pyruvate on hepatic FGF21 expression and secretion remains unknown. Herein, we examined the gene expression and protein levels of FGF21 in human hepatoma HepG2 cells and mouse AML12 hepatocytes in vitro, as well as in mice in vivo. In HepG2 and AML12 cells, pyruvate at concentrations above 0.1 mM significantly increased FGF21 expression and secretion. The increase in cellular cAMP levels by adenylyl cyclase activation, phosphodiesterase (PDE) inhibition and 8-Bromo-cAMP administration significantly restrained pyruvate-stimulated FGF21 expression. Pyruvate significantly increased PDE activities, reduced cAMP levels and decreased CREB phosphorylation. The inhibition of exchange protein directed activated by cAMP (Epac) and cAMP response element binding protein (CREB) upregulated FGF21 expression, upon which pyruvate no longer increased FGF21 expression. The increase in plasma pyruvate levels in mice induced by the intraperitoneal injection of pyruvate significantly increased FGF21 gene expression and PDE activity with a reduction in cAMP levels and CREB phosphorylation in the mouse liver compared with the control. In conclusion, pyruvate activates PDEs to reduce cAMP and then inhibits the cAMP-Epac-CREB signaling pathway to upregulate FGF21 expression in hepatocytes.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Fibroblast Growth Factors , Guanine Nucleotide Exchange Factors , Liver , Phosphoric Diester Hydrolases , Pyruvic Acid , Animals , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Hep G2 Cells , Humans , Liver/enzymology , Liver/metabolism , Mice , Phosphoric Diester Hydrolases/metabolism , Pyruvic Acid/blood , Pyruvic Acid/metabolism , Pyruvic Acid/pharmacokinetics , Signal Transduction/physiology
2.
Can J Physiol Pharmacol ; 100(4): 324-333, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34670103

ABSTRACT

Functional and structural adaptation of common carotid artery could be one of the important causes of postflight orthostatic intolerance after microgravity exposure, the mechanisms of which remain unclear. Recent evidence indicates that long-term spaceflight increases carotid artery stiffness, which might present a high risk to astronaut health and postflight working ability. Studies have suggested that vascular calcification is a common pathological change in cardiovascular diseases that is mainly manifested as an increase in vascular stiffness. Therefore, this study investigated whether simulated microgravity induces calcification of common carotid artery and to elucidate the underlying mechanisms. Four-week-old hindlimb-unweighted (HU) rats were used to simulate the deconditioning effects of microgravity on cardiovascular system. We found that simulated microgravity induced vascular smooth muscle cell (VSMC) osteogenic differentiation and medial calcification, increased receptor activator of nuclear factor κB (NF-κB) ligand (RANKL) and RANK expression, and enhanced NF-κB activation in rat common carotid artery. In vitro activation of the RANK pathway with exogenous RANKL, a RANK ligand, increased RANK and osteoprotegerin (OPG) expression in HU rats. Moreover, the expression of osteogenic markers and activation of NF-κB in HU rats were further enhanced by exogenous RANKL but suppressed by the RANK inhibitor osteoprotegerin fusion protein (OPG-Fc). These results indicated that the OPG/RANKL/RANK system modulates VSMC osteogenic differentiation and medial calcification of common carotid artery in simulated microgravity rats by regulating the NF-kB pathway.


Subject(s)
Osteoprotegerin , Weightlessness , Animals , Carotid Artery, Common/metabolism , NF-kappa B/metabolism , Osteogenesis , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Rats , Weightlessness/adverse effects
3.
Peptides ; 123: 170181, 2020 01.
Article in English | MEDLINE | ID: mdl-31689455

ABSTRACT

Previous studies suggested that endoplasmic reticulum (ER) stress induced-apoptosis promoted vascular calcification (VC). Interestingly, erythropoietin (EPO), an endogenous glycoprotein, exerts multiple tissue protective effects by inhibiting ER stress and apoptosis. We investigated the role and potential mechanism of EPO on VC in chronic kidney disease (CKD) rats and cultured vascular smooth muscle cells (VSMCs). The calcification model was established by subtotal nephrectomy in vivo or phosphate overload in vitro. The protein level of EPO receptor (EPOR) was increased in the calcified aortas of CKD rats. EPO prevented the reduction of VSMC phenotypic markers, and reversed the increased calcium content and calcium salt deposition in the aortas of CKD rats and cultured calcified VSMCs. The protein levels of activating transcription factor 4 (ATF4) and glucose-regulated protein 94 (GRP94) were upregulated in aortas and VSMCs under calcifying conditions, indicating ER stress activation. EPO treatment of CKD rats or calcified VSMCs downregulated the protein levels of ATF4 and GRP94. Furthermore, ER stress-mediated apoptosis, determined by the protein levels of CCAAT/enhancer-binding protein-homologous protein and cleaved caspase 12, was increased in tunicamycin or calcification media-treated VSMCs, but the increased effect was reversed in EPO-treated groups. The increased apoptotic cells in calcified VSMCs, as indicated by Hoechst staining and flow cytometry, were downregulated by the co-administration of EPO or 4-phenyl butyric acid. In conclusion, EPO might attenuate VC by inhibiting ER stress mediated apoptosis through EPOR signaling.


Subject(s)
Aorta/metabolism , Endoplasmic Reticulum Stress , Erythropoietin/metabolism , Renal Insufficiency, Chronic/metabolism , Vascular Calcification/metabolism , Activating Transcription Factor 4/biosynthesis , Animals , Aorta/pathology , Cells, Cultured , Male , Membrane Glycoproteins/biosynthesis , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/pathology , Vascular Calcification/pathology
4.
Cardiorenal Med ; 10(1): 42-50, 2020.
Article in English | MEDLINE | ID: mdl-31694019

ABSTRACT

INTRODUCTION: Vascular calcification (VC) is a complex, regulated process involved in many disease entities. So far, there are no treatments to reverse it. Exploring novel strategies to prevent VC is important and necessary for VC-related disease intervention. OBJECTIVE: In this study, we evaluated whether MOTS-c, a novel mitochondria-related 16-aa peptide, can reduce vitamin D3 and nicotine-induced VC in rats. METHODS: Vitamin D3 plus nicotine-treated rats were injected with MOTS-c at a dose of 5 mg/kg once a day for 4 weeks. Blood pressure, heart rate, and body weight were measured, and echocardiography was performed. The expression of phosphorylated adenosine monophosphate-activated protein kinase (AMPK) and the angiotensin II type 1 (AT-1) and endothelin B (ET-B) receptors was determined by Western blot analysis. RESULTS: Our results showed that MOTS-c treatment significantly attenuated VC. Furthermore, we found that the level of phosphorylated AMPK was increased and the expression levels of the AT-1 and ET-B receptors were decreased after MOTS-c treatment. CONCLUSIONS: Our findings provide evidence that MOTS-c may act as an inhibitor of VC by activating the AMPK signaling pathway and suppressing the expression of the AT-1 and ET-B receptors.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Mitochondrial Proteins/metabolism , Vascular Calcification/metabolism , Animals , Cholecalciferol/administration & dosage , Cholecalciferol/adverse effects , Cholecalciferol/metabolism , Male , Mitochondrial Proteins/administration & dosage , Mitochondrial Proteins/adverse effects , Mitochondrial Proteins/pharmacology , Models, Animal , Nicotine/administration & dosage , Nicotine/adverse effects , Nicotine/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism , Receptor, Endothelin B/drug effects , Receptor, Endothelin B/metabolism , Signal Transduction/drug effects , Vascular Calcification/chemically induced , Ventricular Remodeling/drug effects
5.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 48(5): 716-720, 2017 Sep.
Article in Chinese | MEDLINE | ID: mdl-29130663

ABSTRACT

OBJECTIVE: To investigate the role of capsaicin (CAP) in myocardial ischemia reperfusion injury and its underlying mechanisms. METHODS: Twentyfour adult male SD rats were randomized into 4 groups,namely the control group,ischemia reperfusion group,ischemia reperfusion with CAP group,and ischemia reperfusion with CAPZ and CAP group. Isolated rat hearts underwent Langendorff perfusion. Left ventricular enddiastolic pressure (LVEDP) andleft ventricular developed pressure (LVDP) was calculated to evaluate myocardial performance at 30 min of reperfusion.Triphenyltetrazolium chloride staining was used to measure the infarct size of myocardium at 120 min reperfusion. The morphological changes in myocardial fiber was analyzed by HE staining at the end of reperfusion. Lactate dehydrogenase (LDH) content in the coronary flow was determined during the first 5 min reperfusion. The myocardial mitochondria was isolated and extracted for measuring a series of indicators of mitochondrial oxidative stress,including superoxide dismutase (SOD),methane dicarboxylic aldehyde (MDA) at the end of reperfusion. Western blot was used to determine the expression of caspase3 and cytochrome c at 10 min reperfusion. RESULTS: Compared with the control group,IR group significantly decreased in cardiac function,the level of LVDP and SOD activity and induced an enlarged infarct size ( P<0.01),accompanied by the disordered arrangement of myocardial cells,the content of MDA was increased ( P<0.01),the content of caspase3 and cytochrome c were also upregulated ( P<0.01).10 µmol/L CAP significantly attenuated these effects induced by ischemia reperfusion injury,levels of LVDP and infarct size at the end of reperfusion were significantly improved( P<0.01),nevertheless levels of LVEDP and MDA at the end of reperfusion and LDH were down-regulated markedly ( P<0.01),the content of caspase-3 and cytochrome c were also decreased ( P<0.01). CONCLUSION: These results demonstrate that CAP can suppresses cell apoptosis and necrosis,and alleviate heart function and cell survival from ischemia reperfusion injury through attenuating mitochondrial oxidative stress.


Subject(s)
Capsaicin/pharmacology , Myocardial Reperfusion Injury/drug therapy , Oxidative Stress , Animals , Apoptosis , Caspase 3/metabolism , Cytochromes c/metabolism , Male , Malondialdehyde/metabolism , Myocardium/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
6.
Nutr Metab (Lond) ; 14: 46, 2017.
Article in English | MEDLINE | ID: mdl-28702069

ABSTRACT

BACKGROUND: Musclin is a novel skeletal muscle-derived secretory factor considered to be a potent regulator of the glucose metabolism and therefore may contribute to the pathogenesis of obesity and insulin resistance (IR). METHODS: To test this hypothesis, we examined the plasma musclin levels in overweight/obese subjects and lean controls. Rats on a high fat diet (HFD) were used as the annimal model of obesity. Radioimmunoassay and western blot were used to determine musclin levels in plasma and skeletal muscle. RESULTS: According to radioimmunoassays,the overweight/obese subjects exhibited elevated musclin plasma levels compared with the lean controls (89.49 ± 19.00 ng/L vs 80.39 ± 16.35 ng/L, P < 0.01). The musclin levels were positively correlated with triglyceride, fasting plasma glucose, and homeostasis model assessment of IR levels. These observations were confirmed with a high-fat diet(HFD) rat model. HFD rats also exhibited increased musclin immunoreactivity in plasma (P < 0.01) and in skeletal muscle (P < 0.05), as well as increased musclin mRNA levels in skeletal muscle (P < 0.01). Musclin incubation significantly inhibited muscles 3H-2-DG uptake in the normal diet(ND) group (P < 0.01). The protein expression of glucose transporter type 4 was significantly down regulated by 30% (P < 0.05) in the ND group after soleusmuscle was incubated with musclin compared with the control. Musclin incubation also increased the protein levels of glucose-regulated protein (GRP)78 and GRP94 by 146.8 and 54% (both P < 0.05), respectively, in ND rats. CONCLUSIONS: Our data support the hypothesis that musclin has a strong relationship with obesity-associated IR by impairing the glucose metabolism and, at least in part, through causing endoplasmic reticulum stress.

7.
Arterioscler Thromb Vasc Biol ; 36(11): 2176-2190, 2016 11.
Article in English | MEDLINE | ID: mdl-27634835

ABSTRACT

OBJECTIVE: Oxidative stress plays a critical role in the development of abdominal aortic aneurysm (AAA). Intermedin (IMD) is a regulator of oxidative stress. Here, we investigated whether IMD reduces AAA by inhibiting oxidative stress. APPROACH AND RESULTS: In angiotensin II-induced ApoE-/- mouse and CaCl2-induced C57BL/6J mouse model of AAA, IMD1-53 significantly reduced the incidence of AAA and maximal aortic diameter. Ultrasonography, hematoxylin, and eosin staining and Verhoeff-van Gieson staining showed that IMD1-53 significantly decreased the enlarged aortas and elastic lamina degradation induced by angiotensin II or CaCl2. Mechanistically, IMD1-53 attenuated oxidative stress, inflammation, vascular smooth muscle cell apoptosis, and matrix metalloproteinase activation. IMD1-53 inhibited the activation of redox-sensitive signaling pathways, decreased the mRNA and protein expression of nicotinamide adenine dinucleotide phosphate oxidase subunits, and reduced the activity of nicotinamide adenine dinucleotide phosphate oxidase in AAA mice. Expression of Nox4 was upregulated in human AAA segments and in angiotensin II-treated mouse aortas and was markedly decreased by IMD1-53. In vitro, vascular smooth muscle cells with small-interfering RNA knockdown of IMD showed significantly increased angiotensin II-induced reactive oxygen species, and small-interfering RNA knockdown of Nox4 markedly inhibited the reactive oxygen species. IMD knockdown further increased the apoptosis of vascular smooth muscle cells and inflammation, which was reversed by Nox4 knockdown. Preincubation with IMD17-47 and protein kinase A inhibitor H89 inhibited the effect of IMD1-53, reducing Nox4 protein levels. CONCLUSIONS: IMD1-53 could have a protective effect on AAA by inhibiting oxidative stress.


Subject(s)
Antioxidants/pharmacology , Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/prevention & control , Oxidative Stress/drug effects , Peptide Hormones/pharmacology , Adrenomedullin/metabolism , Angiotensin II , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/drug effects , Calcium Chloride , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dilatation, Pathologic , Disease Models, Animal , Genotype , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NADPH Oxidases/metabolism , Neuropeptides/metabolism , Peptide Hormones/metabolism , Phenotype , RNA Interference , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , Transfection
8.
Kidney Int ; 89(3): 586-600, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26880455

ABSTRACT

Deficiency in α-Klotho is involved in the pathogenesis of vascular calcification. Since intermedin (IMD)1-53 (a calcitonin/calcitonin gene-related peptide) protects against vascular calcification, we studied whether IMD1-53 inhibits vascular calcification by upregulating α-Klotho. A rat model of chronic kidney disease (CKD) with vascular calcification induced by the 5/6 nephrectomy plus vitamin D3 was used for study. The aortas of rats with CKD showed reduced IMD content but an increase of its receptor, calcitonin receptor-like receptor, and its receptor modifier, receptor activity-modifying protein 3. IMD1-53 treatment reduced vascular calcification. The expression of α-Klotho was greatly decreased in the aortas of rats with CKD but increased in the aortas of IMD1-53-treated rats with CKD. In vitro, IMD1-53 increased α-Klotho protein level in calcified vascular smooth muscle cells. α-Klotho knockdown blocked the inhibitory effect of IMD1-53 on vascular smooth muscle cell calcification and their transformation into osteoblast-like cells. The effect of IMD1-53 to upregulate α-Klotho and inhibit vascular smooth muscle cell calcification was abolished by knockdown of its receptor or its modifier protein, or treatment with the protein kinase A inhibitor H89. Thus, IMD1-53 may attenuate vascular calcification by upregulating α-Klotho via the calcitonin receptor/modifying protein complex and protein kinase A signaling.


Subject(s)
Cell Transdifferentiation/drug effects , Glucuronidase/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Osteoblasts/drug effects , Peptide Hormones/pharmacology , Renal Insufficiency, Chronic/drug therapy , Vascular Calcification/prevention & control , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Cells, Cultured , Cholecalciferol , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Glucuronidase/genetics , Humans , Klotho Proteins , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Nephrectomy , Osteoblasts/metabolism , Osteoblasts/pathology , Phenotype , RNA Interference , Rats, Sprague-Dawley , Receptor Activity-Modifying Protein 3/metabolism , Receptors, Calcitonin/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Signal Transduction/drug effects , Transfection , Up-Regulation , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
9.
Sheng Li Ke Xue Jin Zhan ; 46(4): 245-9, 2015 Aug.
Article in Chinese | MEDLINE | ID: mdl-26669073

ABSTRACT

Klotho, a newly identified anti-aging gene, can be regulated by many factors, such as calcitonin gene-related peptide, fibroblast growth factor 2 could up-regulate Klotho expression; whereas renin-angiotensin system, urinary toxins, inflammation and oxidative stress could reduce expression of Klotho. There are two forms of Klotho protein: membrane-bound Klotho and secreted Klotho. Existing studies showed that Klotho was involved in the development of many diseases, including vascular calcification, atherosclerosis, hypertension, kidney damage, hyperparathyroidism, diabetes and tumors. In this paper, the regulation of Klotho expression and its role in diseases are reviewed briefly.


Subject(s)
Glucuronidase/metabolism , Aging , Kidney , Klotho Proteins , Oxidative Stress , Renin-Angiotensin System , Vascular Calcification
10.
Sci Rep ; 5: 14971, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26455818

ABSTRACT

Oxidative stress and inflammation play crucial role in the pathogenesis of chronic obstructive pulmonary disease (COPD). Most patients with COPD show a poor response to corticosteroids. Hydrogen sulfide (H2S ) has been implicated in the pathogenesis of COPD, but its expression and effects in lung tissue from COPD patients are not clear. In peripheral lung tissue samples from 24 patients, we found that compared with nonsmokers, the protein level of cystathionine-γ-lyase (CSE) was decreased in smokers and COPD patients. CSE mRNA increased but cystathionine-ß-synthase (CBS) mRNA decreased in COPD patients. H2S donors increased glutathione and superoxide dismutase in CS exposed U937 cells and inhibited CS-induced TNF-α and IL-8 secretion. Dexamethasone alone had no effect on lipopolysaccharide (LPS) induced TNF-α release by alveolar macrophages from CS exposed rats, however the combination of dexamethasone and H2S donor significantly inhibited TNF-α release. Thus, H2S metabolism is altered in lung tissue of smokers and COPD patients. Supplementation of H2S protects against CS-induced oxidative stress and inflammation in macrophages and H2S on steroid sensitivity deserves further investigation.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Anti-Inflammatory Agents/pharmacology , Lung/metabolism , Macrophages/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , RNA, Messenger/metabolism , Animals , Cell Line, Tumor , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation , Glutathione/metabolism , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Interleukin-8/genetics , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/pathology , Macrophages/drug effects , Macrophages/pathology , Oxidative Stress , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction , Smoking/adverse effects , Sulfides/metabolism , Sulfides/pharmacology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Heart Vessels ; 30(5): 657-68, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25092223

ABSTRACT

Fibroblast growth factor 21 (FGF-21) is an endocrine factor that can be secreted into circulation by the liver. FGF-21 takes part in metabolic actions and is thought to be a promising candidate for the treatment of diabetes. However, the role of FGF-21 in atherosclerosis is unknown. In this study, apoE(-/-) mice were fed an atherogenic diet for 4 weeks with and without subcutaneous injections of FGF-21. ApoE(-/-) mice fed an atherogenic diet showed hyperlipidemia, a large plaque area in aortas and increased vessel wall thickness. Plasma FGF-21 content and protein level of FGF receptor 1 (FGFR1) in aortas was greater in apoE(-/-) than C57BL/6J mice. Exogenous FGF-21 treatment significantly ameliorated dyslipidemia in apoE(-/-) mice. FGF-21-treated apoE(-/-) mice showed reduced number of aortic plaques and plaque area as well as reduced number of TUNEL-positive cells. Protein levels of the endoplasmic reticulum stress markers glucose-regulated protein 94, caspase-12 and C/EBP homologous protein were reduced by 34.5, 31.4 and 26.5 %, respectively, in apoE(-/-) mice. Endogenous expression of FGF-21 and its receptor FGFR1 were upregulated in apoE(-/-) mice, and exogenous administration of FGF-21 ameliorated the atherogenic-induced dyslipidemia and vascular atherosclerotic lesions. FGF-21 protecting against atherosclerosis might be in part by its inhibitory effects on endoplasmic reticulum stress-mediated apoptosis.


Subject(s)
Apolipoproteins E/deficiency , Apoptosis , Atherosclerosis/drug therapy , Dyslipidemias/drug therapy , Endoplasmic Reticulum Stress/drug effects , Fibroblast Growth Factors/biosynthesis , Animals , Apolipoproteins E/blood , Atherosclerosis/etiology , Atherosclerosis/pathology , Blotting, Western , Disease Models, Animal , Dyslipidemias/complications , Dyslipidemias/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/therapeutic use , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Radioimmunoassay
13.
Calcif Tissue Int ; 96(1): 80-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25416842

ABSTRACT

Thyroid hormones (THs) including thyroxine (T4) and triiodothyronine (T3) play critical roles in bone remodeling. However, the role and mechanism of THs in vascular calcification (VC) have been unclear. To explore the pathophysiological roles of T3 on VC, we investigated the changes in plasma and aortas of THs concentrations and the effect of T3 on rat VC induced by vitamin D3 plus nicotine (VDN). VDN-treated rat showed decreased plasma T3 content, increased vascular calcium deposition, and alkaline phosphatase (ALP) activity. Administration of T3 (0.2 mg/kg body weight IP) for 10 days greatly reduced vascular calcium deposition and ALP activity in calcified rat aortas when compared with controls. Concurrently, the loss of smooth muscle lineage markers α-actin and SM22a was restored, and the increased bone-associated molecules, such as runt-related transcription factor2 (Runx2), Osterix, and osteopontin (OPN) levels in calcified aorta, were reduced by administration of T3. The suppression of klotho in calcified rat aorta was restored by T3. Methimazole (400 mg/L) blocked the beneficial effect of T3 on VC. These results suggested that T3 can inhibit VC development.


Subject(s)
Bone Remodeling/drug effects , Cholecalciferol/pharmacology , Nicotine/pharmacology , Thyroid Hormones/pharmacology , Vascular Calcification/drug therapy , Animals , Bone and Bones/blood supply , Disease Models, Animal , Male , Osteopontin/metabolism , Rats, Sprague-Dawley , Thyroid Hormones/metabolism , Vascular Calcification/chemically induced
14.
Exp Biol Med (Maywood) ; 239(10): 1352-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24872434

ABSTRACT

Intermedin (IMD) is a member of calcitonin/calcitonin gene-related peptide (CGRP) and involves in the regulation of cardiovascular function in both peripheral tissues and central nervous system (CNS). Paraventricular nucleus (PVN) of hypothalamus is an important site in the control of cardiac sympathetic afferent reflex (CSAR) which participates in sympathetic over-excitation of hypertension. The aim of this study is to investigate whether IMD in the PVN is involved in the inhibition of CSAR and its related mechanism in hypertension. Rats were subjected to two-kidney one-clip (2K1C) surgery to induce renovascular hypertension or sham-operation (Sham). Acute experiments were carried out four weeks later under anesthesia. The CSAR was evaluated with the renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) responses to the epicardial application of capsaicin. The RSNA and MAP were recorded in sinoaortic-denervated, cervical-vagotomized and anesthetized rats. Bilateral PVN microinjection of IMD (25 pmol) caused greater decrease in the CSAR in 2K1C rats than in Sham rats, which was prevented by pretreatment with adrenomedullin (AM) receptor antagonist AM22-52, non-selective nitric oxide (NO) synthase (NOS) inhibitor L-NAME or γ-amino butyric acid (GABA)B receptor blocker CGP-35348. PVN pretreatment with CGRP receptor antagonist CGRP8-37 or GABA(A) receptor blocker gabazine had no significant effect on the CSAR response to IMD. AM22-52, L-NAME and CGP-35348 in the PVN could increase CSAR in Sham and 2K1C rats. These data indicate that IMD in the PVN inhibits CSAR via AM receptor, and both NO and GABA in the PVN involve in the effect of IMD on CSAR in Sham and renovascular hypertensive rats.


Subject(s)
Adrenomedullin/administration & dosage , Afferent Pathways/drug effects , Neuropeptides/administration & dosage , Nitric Oxide/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Reflex , Sympathetic Nervous System/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Male , Microinjections/methods , Rats, Sprague-Dawley
15.
Exp Biol Med (Maywood) ; 238(10): 1136-46, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24006303

ABSTRACT

We previously reported that endoplasmic reticulum (ER) stress-mediated apoptosis participated in vascular calcification. Importantly, a novel paracrine/autocrine peptide intermedin1-53 (IMD1-53) in the vasculature inhibited vascular calcification in rats. But the mechanisms needed to be fully elucidated. Vascular smooth muscle cells (VSMCs) calcification was induced by CaCl2 and ß-glycerophosphate. Tunicamycin (Tm) or dithiothreitol (DTT) was used to induce ER stress. We found that IMD1-53 (10(-7)mol/L) treatment significantly alleviated the protein expression of ER stress hallmarks activating transcription factor 4 (ATF4), ATF6, glucose-regulated protein 78 (GRP78) and GRP94 induced by Tm or DTT. ER stress occurred in early and late calcification of VSMCs but was inhibited by IMD1-53. These inhibitory effects of IMD1-53 were abolished by treatment with the protein kinase A (PKA) inhibitor H89. Pretreatment with IMD1-53 decreased the number of apoptotic VSMCs and downregulated protein expression of cleaved caspase 12 and C/EBP homologous protein (CHOP) in calcified VSMCs. Concurrently, IMD1-53 restored the loss of VSMC lineage markers and ameliorated calcium deposition and alkaline phosphatase activity in calcified VSMCs as well. The observation was further verified by Alizarin Red S staining, which showed that IMD1-53 reduced positive red nodules among calcified VSMCs. In conclusion, IMD1-53 attenuated VSMC calcification by inhibiting ER stress through cAMP/PKA signalling.


Subject(s)
Adrenomedullin/metabolism , Calcinosis/physiopathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Endoplasmic Reticulum Stress/drug effects , Muscle, Smooth, Vascular/drug effects , Neuropeptides/metabolism , Vascular Calcification/physiopathology , Animals , Apoptosis/drug effects , Cells, Cultured , Male , Muscle, Smooth, Vascular/physiopathology , Rats , Rats, Sprague-Dawley , Signal Transduction
16.
Peptides ; 48: 156-65, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23994559

ABSTRACT

Ghrelin, the endogenous ligand of growth hormone secretagogue receptor (GHS-R), is a cardioprotective peptide. In our previous work, we have revealed that ghrelin could protect heart against ischemia/reperfusion (I/R) injury by inhibiting endoplasmic reticulum stress (ERS), which contributes to many heart diseases. In current study, using both in vivo and in vitro models, we investigated how ghrelin inhibits myocardial ERS. In the in vivo rat heart injury model induced by isoproterenol (ISO), we found that exogenous ghrelin could alleviate heart dysfunction, reduce myocardial injury and apoptosis and inhibit the excessive myocardial ERS induced by ISO. More importantly, the activation of AMP-activated protein kinase (AMPK) was observed. To explore the role of AMPK activation in ERS inhibition by ghrelin, we set up two in vitro ERS models by exposing cultured rat cardiomyocytes to tunicamycin(Tm) or dithiothreitol (DTT). In both models, compared with Tm or DTT treatment alone, pre-incubation cardiomyocytes with ghrelin significantly activated AMPK, reversed the upregulation of the ERS markers, C/EBP-homologous protein (CHOP) and cleaved caspase-12, and reduced apoptosis of cardiomyocytes. Further, we found that the ERS inhibitory and anti-apoptotic actions induced by ghrelin were blocked by an AMPK inhibitor. To investigate how ghrelin activates AMPK, selective antagonist of GHS-R1a and inhibitor of Ca(2+)/Calmodulin-dependent protein kinase kinase (CaMKK) were added, respectively, before ghrelin pre-incubation, and we found that AMPK activation was prevented and the ERS inhibitory and anti-apoptotic actions of ghrelin were blocked. In conclusion, ghrelin could protect heart against ERS-induced injury and apoptosis, at least partially through a GHS-R1a/CaMKK/AMPK pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cardiotonic Agents/pharmacology , Ghrelin/pharmacology , Heart Injuries/drug therapy , Animals , Apoptosis/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Ghrelin/metabolism , Heart Injuries/chemically induced , Humans , Isoproterenol/toxicity , Metabolic Networks and Pathways/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Receptors, Ghrelin/metabolism , Reperfusion Injury/chemically induced , Reperfusion Injury/drug therapy , Tunicamycin/pharmacology
17.
Apoptosis ; 18(9): 1132-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23686245

ABSTRACT

Our previous work reported that endoplasmic reticulum stress (ERS)-mediated apoptosis was activated during vascular calcification (VC). Activating transcription factor 4 (ATF4) is a critical transcription factor in osteoblastogenesis and ERS-induced apoptosis. However, whether ATF4 is involved in ERS-mediated apoptosis contributing to VC remains unclear. In the present study, in vivo VC was induced in rats by administering vitamin D3 plus nicotine. Vascular smooth muscle cell (VSMC) calcification in vitro was induced by incubation in calcifying media containing ß-glycerophosphate and CaCl2. ERS inhibitors taurine or 4-phenylbutyric acid attenuated ERS and VSMC apoptosis in calcified rat arteries, reduced calcification and retarded the VSMC contractile phenotype transforming into an osteoblast-like phenotype in vivo. Inhibition of ERS retarded the VSMC phenotypic transition into an osteoblast-like cell phenotype and reduced VSMC calcification and apoptosis in vitro. Interestingly, ATF4 was activated in calcified aortas and calcified VSMCs in vitro. ATF4 knockdown attenuated ERS-induced apoptosis in calcified VSMCs. ATF4 deficiency blocked VSMC calcification and negatively regulated the osteoblast phenotypic transition of VSMCs in vitro. Our results demonstrate that ATF4 was involved at least in part in the process of ERS-mediated apoptosis contributing to VC.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis , Endoplasmic Reticulum Stress , Muscle, Smooth, Vascular/cytology , Vascular Calcification/metabolism , Vascular Calcification/physiopathology , Activating Transcription Factor 4/genetics , Animals , Cells, Cultured , Humans , Male , Muscle, Smooth, Vascular/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Rats , Rats, Sprague-Dawley , Vascular Calcification/genetics
18.
Peptides ; 42: 25-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23291307

ABSTRACT

Angiotensin-(1-7) [Ang-(1-7)] is a new bioactive heptapeptide in the renin-angiotensin-aldosterone system (RAAS) with potent protective effects in cardiovascular diseases, opposing many actions of angiotensin II (Ang II) mediated by Ang II type 1 (AT1) receptor. It is produced mainly by the activity of angiotensin-converting enzyme 2 (ACE2) and acts through the Mas receptor. However, the role of Ang-(1-7) in vascular calcification (VC) is still unclear. In this study, we investigated the protective effects of Ang-(1-7) on VC in an in vivo rat VC model induced by vitamin D3 plus nicotine. The levels of ACE2 and the Mas receptor, as well as ACE, AT1 receptor, Ang II type 2 receptor and angiotensinogen, were significantly increased in calcified aortas, and Ang-(1-7) reversed the increased levels. Ang-(1-7) restored the reduced expression of lineage markers, including smooth muscle (SM) α-actin, SM22α, calponin and smoothelin, in vascular smooth muscle cells (VSMCs) and retarded the osteogenic transition of VSMCs by decreasing the expression of bone-associated proteins. It reduced alkaline phosphatase activity and calcium deposition in VC and alleviated the hemodynamic disorders of rats with VC. We provide the first in vivo evidence that Ang-(1-7) can inhibit the development of VC by inhibiting the osteogenic transition of VSMCs, at least in part by decreasing levels of the ACE/Ang II/AT1 axis. The increased expression of ACE2 and the Mas receptor in calcified aortas suggests the involvement of the ACE2/Ang-(1-7)/Mas axis during VC. Ang-(1-7) might be an efficient endogenous vasoprotective factor for VC.


Subject(s)
Angiotensin I/pharmacology , Peptide Fragments/pharmacology , Vascular Calcification/drug therapy , Vascular Calcification/metabolism , Actins/genetics , Alkaline Phosphatase/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Biomarkers/metabolism , Calcium-Binding Proteins/genetics , Cholecalciferol/adverse effects , Cytoskeletal Proteins/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Male , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nicotine/toxicity , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Vascular Calcification/chemically induced , Vascular Calcification/genetics , Calponins
19.
Cell Biochem Biophys ; 66(3): 421-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23274912

ABSTRACT

Peroxisome proliferator-activated receptor γ (PPARγ) and its ligands have profound effects on glucose homeostasis, cardiovascular diseases, and bone metabolism. To explore the pathophysiological roles of PPARγ in diabetes with concomitant vascular calcification, we investigated changes in PPARγ expression and the effect of the PPARγ ligands troglitazone and rosiglitazone on vascular smooth muscle cell (VSMC) calcification induced by high glucose (HG, 25 mmol/L). Compared with low glucose, HG-induced VSMC calcification, and PPARγ mRNA, protein level was decreased. Troglitazone and rosiglitazone treatment markedly attenuated the VSMC calcification, whereas PPARγ antagonist GW9662 abolished the effect of rosiglitazone on calcification. Pretreatment of VSMCs with rosiglitazone, but not troglitazone, restored the loss of lineage marker expression: the protein levels of α-actin and SM-22α were increased 52 % (P < 0.05) and 53.1% (P < 0.01), respectively, as compared with HG alone. Troglitazone and rosiglitazone reversed the change in bone-related protein expression induced by HG: decreased the mRNA levels of osteocalcin, bone morphogenetic protein 2 (BMP2), and core binding factor α 1 (Cbfα-1) by 26.9% (P > 0.05), 50.0 % (P < 0.01), and 24.4% (P < 0.05), and 48.4% (P < 0.05), 41.4% (P < 0.01) and 56.2% (P < 0.05), respectively, and increased that of matrix Gla protein (MGP) 84.2% (P < 0.01) and 70.0%, respectively (P < 0.05), as compared with HG alone. GW9662 abolished the effect of rosiglitazone on Cbfα-1 and MGP expression. PPARγ ligands can inhibit VSMCs calcification induced by high glucose.


Subject(s)
Calcinosis/chemically induced , Calcinosis/prevention & control , Glucose/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , PPAR gamma/metabolism , Anilides/pharmacology , Animals , Biomarkers/metabolism , Calcinosis/metabolism , Calcinosis/pathology , Cells, Cultured , Chromans/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Ligands , Male , Muscle, Smooth, Vascular/cytology , Osteoblasts/cytology , Osteoblasts/drug effects , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Rosiglitazone , Thiazolidinediones/pharmacology , Troglitazone , Vasoconstriction/drug effects
20.
Sheng Li Ke Xue Jin Zhan ; 41(4): 248-55, 2010 Aug.
Article in Chinese | MEDLINE | ID: mdl-21416939

ABSTRACT

Skeleton has long been recognized as a organ supporting body system and regulating metabolism of calcium and phosphorus. Recent researches found that bone cells, especially osteoblast and osteoclast, synthesize and secrete various bioactive molecules, such as bone morphogenetic proteins, growth factors, adipokines, inflammatory cytokines and cardiovascular bioactive peptides. The active factors produced by bone not only play important roles in the skeleton system per se by paracrine/autocrine pathway but also regulate energy metabolism, inflammatory process, endocrine homeostasis by endocrine pathway.


Subject(s)
Bone and Bones/metabolism , Bone and Bones/physiology , Osteoblasts/metabolism , Osteoclasts/metabolism , Bone Morphogenetic Proteins/metabolism , Endocrine System/metabolism , Humans , Osteoblasts/physiology , Osteocalcin/metabolism , Osteoclasts/physiology , Osteoprotegerin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...